38 results on '"Florian Giesert"'
Search Results
2. Engineered, nucleocytoplasmic shuttling Cas13d enables highly efficient cytosolic RNA targeting
- Author
-
Christoph Gruber, Lea Krautner, Valter Bergant, Vincent Grass, Zhe Ma, Lara Rheinemann, Ariane Krus, Friederike Reinhardt, Lyupka Mazneykova, Marianne Rocha-Hasler, Dong-Jiunn Jeffery Truong, Gil Gregor Westmeyer, Andreas Pichlmair, Gregor Ebert, Florian Giesert, and Wolfgang Wurst
- Subjects
Cytology ,QH573-671 - Published
- 2024
- Full Text
- View/download PDF
3. The thymocyte-specific RNA-binding protein Arpp21 provides TCR repertoire diversity by binding to the 3’-UTR and promoting Rag1 mRNA expression
- Author
-
Meng Xu, Taku Ito-Kureha, Hyun-Seo Kang, Aleksandar Chernev, Timsse Raj, Kai P. Hoefig, Christine Hohn, Florian Giesert, Yinhu Wang, Wenliang Pan, Natalia Ziętara, Tobias Straub, Regina Feederle, Carolin Daniel, Barbara Adler, Julian König, Stefan Feske, George C. Tsokos, Wolfgang Wurst, Henning Urlaub, Michael Sattler, Jan Kisielow, F. Gregory Wulczyn, Marcin Łyszkiewicz, and Vigo Heissmeyer
- Subjects
Science - Abstract
Abstract The regulation of thymocyte development by RNA-binding proteins (RBPs) is largely unexplored. We identify 642 RBPs in the thymus and focus on Arpp21, which shows selective and dynamic expression in early thymocytes. Arpp21 is downregulated in response to T cell receptor (TCR) and Ca2+ signals. Downregulation requires Stim1/Stim2 and CaMK4 expression and involves Arpp21 protein phosphorylation, polyubiquitination and proteasomal degradation. Arpp21 directly binds RNA through its R3H domain, with a preference for uridine-rich motifs, promoting the expression of target mRNAs. Analysis of the Arpp21–bound transcriptome reveals strong interactions with the Rag1 3′-UTR. Arpp21–deficient thymocytes show reduced Rag1 expression, delayed TCR rearrangement and a less diverse TCR repertoire. This phenotype is recapitulated in Rag1 3′-UTR mutant mice harboring a deletion of the Arpp21 response region. These findings show how thymocyte-specific Arpp21 promotes Rag1 expression to enable TCR repertoire diversity until signals from the TCR terminate Arpp21 and Rag1 activities.
- Published
- 2024
- Full Text
- View/download PDF
4. Engrailed 1 deficiency induces changes in ciliogenesis during human neuronal differentiation
- Author
-
Sina Hembach, Sebastian Schmidt, Tanja Orschmann, Ingo Burtscher, Heiko Lickert, Florian Giesert, Daniela Vogt Weisenhorn, and Wolfgang Wurst
- Subjects
Engrailed 1 ,dopaminergic neuron ,primary cilia ,Wnt signaling ,induced pluripotent stem cells ,complex I ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
A key pathological feature of Parkinson’s Disease (PD) is the progressive degeneration of dopaminergic neurons (DAns) in the substantia nigra pars compacta. Considering the major role of EN1 in the development and maintenance of these DAns and the implications from En1 mouse models, it is highly interesting to study the molecular and protective effect of EN1 also in a human cellular model. Therefore, we generated EN1 knock-out (ko) human induced pluripotent stem cell (hiPSCs) lines and analyzed these during neuronal differentiation. Although the EN1 ko didn’t interfere with neuronal differentiation and generation of tyrosine hydroxylase positive (TH+) neurons per se, the neurons exhibited shorter neurites. Furthermore, mitochondrial respiration, as well as mitochondrial complex I abundance was significantly reduced in fully differentiated neurons. To understand the implications of an EN1 ko during differentiation, we performed a transcriptome analysis of human neuronal precursor cells (hNPCs) which unveiled alterations in cilia-associated pathways. Further analysis of ciliary morphology revealed an elongation of primary cilia in EN1-deficient hNPCs. Besides, also Wnt signaling pathways were severely affected. Upon stimulating hNPCs with Wnt which drastically increased EN1 expression in WT lines, the phenotypes concerning mitochondrial function and cilia were exacerbated in EN1 ko hNPCs. They failed to enhance the expression of the complex I subunits NDUFS1 and 3, and now displayed a reduced mitochondrial respiration. Furthermore, Wnt stimulation decreased ciliogenesis in EN1 ko hNPCs but increased ciliary length even further. This further highlights the relevance of primary cilia next to mitochondria for the functionality and correct maintenance of human DAns and provides new possibilities to establish neuroprotective therapies for PD.
- Published
- 2024
- Full Text
- View/download PDF
5. A reversible state of hypometabolism in a human cellular model of sporadic Parkinson’s disease
- Author
-
Sebastian Schmidt, Constantin Stautner, Duc Tung Vu, Alexander Heinz, Martin Regensburger, Ozge Karayel, Dietrich Trümbach, Anna Artati, Sabine Kaltenhäuser, Mohamed Zakaria Nassef, Sina Hembach, Letyfee Steinert, Beate Winner, Winkler Jürgen, Martin Jastroch, Malte D. Luecken, Fabian J. Theis, Gil Gregor Westmeyer, Jerzy Adamski, Matthias Mann, Karsten Hiller, Florian Giesert, Daniela M. Vogt Weisenhorn, and Wolfgang Wurst
- Subjects
Science - Abstract
Abstract Sporadic Parkinson’s Disease (sPD) is a progressive neurodegenerative disorder caused by multiple genetic and environmental factors. Mitochondrial dysfunction is one contributing factor, but its role at different stages of disease progression is not fully understood. Here, we showed that neural precursor cells and dopaminergic neurons derived from induced pluripotent stem cells (hiPSCs) from sPD patients exhibited a hypometabolism. Further analysis based on transcriptomics, proteomics, and metabolomics identified the citric acid cycle, specifically the α-ketoglutarate dehydrogenase complex (OGDHC), as bottleneck in sPD metabolism. A follow-up study of the patients approximately 10 years after initial biopsy demonstrated a correlation between OGDHC activity in our cellular model and the disease progression. In addition, the alterations in cellular metabolism observed in our cellular model were restored by interfering with the enhanced SHH signal transduction in sPD. Thus, inhibiting overactive SHH signaling may have potential as neuroprotective therapy during early stages of sPD.
- Published
- 2023
- Full Text
- View/download PDF
6. Toxicity of extracellular alpha-synuclein is independent of intracellular alpha-synuclein
- Author
-
Yanina Dening, Theresa Straßl, Viktoria Ruf, Petra Dirscherl, Alexandra Chovsepian, Alicia Stievenard, Amit Khairnar, Felix Schmidt, Florian Giesert, Jochen Herms, Johannes Levin, Marianne Dieterich, Peter Falkai, Daniela Vogt Weisenhorn, Wolfgang Wurst, Armin Giese, and Francisco Pan-Montojo
- Subjects
Medicine ,Science - Abstract
Abstract Parkinson´s disease (PD) pathology progresses throughout the nervous system. Whereas motor symptoms are always present, there is a high variability in the prevalence of non-motor symptoms. It has been postulated that the progression of the pathology is based on a prion-like disease mechanism partly due to the seeding effect of endocytosed-alpha-synuclein (ASYN) on the endogenous ASYN. Here, we analyzed the role of endogenous ASYN in the progression of PD-like pathology in vivo and in vitro and compared the effect of endocytosed-ASYN as well as paraquat and rotenone on primary enteric, dopaminergic and cortical neurons from wild-type and ASYN-KO mice. Our results show that, in vivo, pathology progression did not occur in the absence of endogenous ASYN. Remarkably, the damage caused by endocytosed-ASYN, rotenone or paraquat was independent from endogenous ASYN and related to the alteration of the host´s mitochondrial membrane potential. Dopaminergic neurons were very sensitive to these noxae compared to other neuronal subtypes. These results suggest that ASYN-mitochondrial interactions play a major role in initiating the pathological process in the host neuron and endogenous ASYN is essential for the transsynaptical transmission of the pathology. Our results also suggest that protecting mitochondrial function is a valid primary therapeutic target.
- Published
- 2022
- Full Text
- View/download PDF
7. Primary cilia and SHH signaling impairments in human and mouse models of Parkinson’s disease
- Author
-
Sebastian Schmidt, Malte D. Luecken, Dietrich Trümbach, Sina Hembach, Kristina M. Niedermeier, Nicole Wenck, Klaus Pflügler, Constantin Stautner, Anika Böttcher, Heiko Lickert, Ciro Ramirez-Suastegui, Ruhel Ahmad, Michael J. Ziller, Julia C. Fitzgerald, Viktoria Ruf, Wilma D. J. van de Berg, Allert J. Jonker, Thomas Gasser, Beate Winner, Jürgen Winkler, Daniela M. Vogt Weisenhorn, Florian Giesert, Fabian J. Theis, and Wolfgang Wurst
- Subjects
Science - Abstract
Here, the authors reveal using single-cell RNA sequencing that Parkinson’s disease (PD) patient-derived neuronal cells show altered primary cilia morphology and signaling suggesting cilia dysfunction may underlie PD pathogenesis.
- Published
- 2022
- Full Text
- View/download PDF
8. Parkinson's disease motor symptoms rescue by CRISPRa‐reprogramming astrocytes into GABAergic neurons
- Author
-
Jessica Giehrl‐Schwab, Florian Giesert, Benedict Rauser, Chu Lan Lao, Sina Hembach, Sandrine Lefort, Ignacio L Ibarra, Christina Koupourtidou, Malte Daniel Luecken, Dong‐Jiunn Jeffery Truong, Judith Fischer‐Sternjak, Giacomo Masserdotti, Nilima Prakash, Jovica Ninkovic, Sabine M Hölter, Daniela M Vogt Weisenhorn, Fabian J Theis, Magdalena Götz, and Wolfgang Wurst
- Subjects
astrocytes ,CRISPRa ,GABAergic neurons ,Parkinson's disease ,reprogramming ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Direct reprogramming based on genetic factors resembles a promising strategy to replace lost cells in degenerative diseases such as Parkinson's disease. For this, we developed a knock‐in mouse line carrying a dual dCas9 transactivator system (dCAM) allowing the conditional in vivo activation of endogenous genes. To enable a translational application, we additionally established an AAV‐based strategy carrying intein‐split‐dCas9 in combination with activators (AAV‐dCAS). Both approaches were successful in reprogramming striatal astrocytes into induced GABAergic neurons confirmed by single‐cell transcriptome analysis of reprogrammed neurons in vivo. These GABAergic neurons functionally integrate into striatal circuits, alleviating voluntary motor behavior aspects in a 6‐OHDA Parkinson's disease model. Our results suggest a novel intervention strategy beyond the restoration of dopamine levels. Thus, the AAV‐dCAS approach might enable an alternative route for clinical therapies of Parkinson's disease.
- Published
- 2022
- Full Text
- View/download PDF
9. Meis1: effects on motor phenotypes and the sensorimotor system in mice
- Author
-
Aaro V. Salminen, Lillian Garrett, Barbara Schormair, Jan Rozman, Florian Giesert, Kristina M. Niedermeier, Lore Becker, Birgit Rathkolb, Ildikó Rácz, German Mouse Clinic Consortium, Martin Klingenspor, Thomas Klopstock, Eckhard Wolf, Andreas Zimmer, Valérie Gailus-Durner, Miguel Torres, Helmut Fuchs, Martin Hrabě de Angelis, Wolfgang Wurst, Sabine M. Hölter, and Juliane Winkelmann
- Subjects
Meis1 ,Prepulse inhibition ,Restless legs syndrome ,Sensorimotor system ,Mouse model ,Pramipexole ,Medicine ,Pathology ,RB1-214 - Abstract
MEIS1 encodes a developmental transcription factor and has been linked to restless legs syndrome (RLS) in genome-wide association studies. RLS is a movement disorder leading to severe sleep reduction and has a substantial impact on the quality of life of patients. In genome-wide association studies, MEIS1 has consistently been the gene with the highest effect size and functional studies suggest a disease-relevant downregulation. Therefore, haploinsufficiency of Meis1 could be the system with the most potential for modeling RLS in animals. We used heterozygous Meis1-knockout mice to study the effects of Meis1 haploinsufficiency on mouse behavioral and neurological phenotypes, and to relate the findings to human RLS. We exposed the Meis1-deficient mice to assays of motor, sensorimotor and cognitive ability, and assessed the effect of a dopaminergic receptor 2/3 agonist commonly used in the treatment of RLS. The mutant mice showed a pattern of circadian hyperactivity, which is compatible with human RLS. Moreover, we discovered a replicable prepulse inhibition (PPI) deficit in the Meis1-deficient animals. In addition, these mice were hyposensitive to the PPI-reducing effect of the dopaminergic receptor agonist, highlighting a role of Meis1 in the dopaminergic system. Other reported phenotypes include enhanced social recognition at an older age that was not related to alterations in adult olfactory bulb neurogenesis previously shown to be implicated in this behavior. In conclusion, the Meis1-deficient mice fulfill some of the hallmarks of an RLS animal model, and revealed the role of Meis1 in sensorimotor gating and in the dopaminergic systems modulating it.
- Published
- 2017
- Full Text
- View/download PDF
10. FGF/FGFR2 signaling regulates the generation and correct positioning of Bergmann glia cells in the developing mouse cerebellum.
- Author
-
Florian Meier, Florian Giesert, Sabit Delic, Theresa Faus-Kessler, Friederike Matheus, Antonio Simeone, Sabine M Hölter, Ralf Kühn, Daniela M Vogt Weisenhorn, Wolfgang Wurst, and Nilima Prakash
- Subjects
Medicine ,Science - Abstract
The normal cellular organization and layering of the vertebrate cerebellum is established during embryonic and early postnatal development by the interplay of a complex array of genetic and signaling pathways. Disruption of these processes and of the proper layering of the cerebellum usually leads to ataxic behaviors. Here, we analyzed the relative contribution of Fibroblast growth factor receptor 2 (FGFR2)-mediated signaling to cerebellar development in conditional Fgfr2 single mutant mice. We show that during embryonic mouse development, Fgfr2 expression is higher in the anterior cerebellar primordium and excluded from the proliferative ventricular neuroepithelium. Consistent with this finding, conditional Fgfr2 single mutant mice display the most prominent defects in the anterior lobules of the adult cerebellum. In this context, FGFR2-mediated signaling is required for the proper generation of Bergmann glia cells and the correct positioning of these cells within the Purkinje cell layer, and for cell survival in the developing cerebellar primordium. Using cerebellar microexplant cultures treated with an FGFR agonist (FGF9) or antagonist (SU5402), we also show that FGF9/FGFR-mediated signaling inhibits the outward migration of radial glia and Bergmann glia precursors and cells, and might thus act as a positioning cue for these cells. Altogether, our findings reveal the specific functions of the FGFR2-mediated signaling pathway in the generation and positioning of Bergmann glia cells during cerebellar development in the mouse.
- Published
- 2014
- Full Text
- View/download PDF
11. Expression analysis of Lrrk1, Lrrk2 and Lrrk2 splice variants in mice.
- Author
-
Florian Giesert, Andreas Hofmann, Alexander Bürger, Julia Zerle, Karina Kloos, Ulrich Hafen, Luise Ernst, Jingzhong Zhang, Daniela Maria Vogt-Weisenhorn, and Wolfgang Wurst
- Subjects
Medicine ,Science - Abstract
Missense mutations in the leucine-rich repeat kinase 2 gene (LRRK2) are linked to autosomal dominant forms of Parkinson's disease (PD). In order to get insights into the physiological role of Lrrk2, we examined the distribution of Lrrk2 mRNA and different splice variants in the developing murine embryo and the adult brain of Mus musculus. To analyse if the Lrrk2-paralog, Lrrk1, may have redundant functions in PD-development, we also compared Lrrk1 and Lrrk2 expression in the same tissues. Using radioactive in situ hybridization, we found ubiquitous expression of both genes at low level from embryonic stage E9.5 onward, which progressively increased up until birth. The developing central nervous system (CNS) displayed no prominent Lrrk2 mRNA signals at these time-points. However, in the entire postnatal brain Lrrk2 became detectable, showing strongest level in the striatum and the cortex of adult mice; Lrrk1 was only detectable in the mitral cell layer of the olfactory bulb. Thus, due to the non-overlapping expression patterns, a redundant function of Lrrk2 and Lrrk1 in the pathogenesis of PD seems to be unlikely. Quantification of Lrrk2 mRNA and protein level in several brain regions by real-time PCR and Western blot verified the striatum and cortex as hotspots of postnatal Lrrk2 expression. Strong expression of Lrrk2 is mainly found in neurons, specifically in the dopamine receptor 1 (DRD1a) and 2 (DRD2)-positive subpopulations of the striatal medium spiny neurons. Finally, we identified 2 new splice-variants of Lrrk2 in RNA-samples from various adult brain regions and organs: a variant with a skipped exon 5 and a truncated variant terminating in an alternative exon 42a. In order to identify the origin of these two splice variants, we also analysed primary neural cultures independently and found cell-specific expression patterns for these variants in microglia and astrocytes.
- Published
- 2013
- Full Text
- View/download PDF
12. Mammalian VPS45 orchestrates trafficking through the endosomal system
- Author
-
Christoph Klein, Eckhard Wolf, Maik Dahlhoff, Florian Giesert, Wolfgang Wurst, Marlon R. Schneider, Benjamin Marquardt, Monika I. Linder, Raz Somech, Laura Frey, Yanshan Liu, Yoko Mizoguchi, Marcin Łyszkiewicz, and Natalia Ziętara
- Subjects
0301 basic medicine ,Endosome ,Immunology ,Vesicular Transport Proteins ,Regulator ,Context (language use) ,Endosomes ,Biology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Humans ,Receptor ,Mice, Knockout ,Vacuolar protein sorting ,Cell Biology ,Hematology ,Cell biology ,Protein Transport ,Haematopoiesis ,030104 developmental biology ,030220 oncology & carcinogenesis ,Lysosomes ,Gene Deletion ,Intracellular ,VPS45 ,HeLa Cells - Abstract
Vacuolar protein sorting 45 homolog (VPS45), a member of the Sec1/Munc18 (SM) family, has been implicated in the regulation of endosomal trafficking. VPS45 deficiency in human patients results in congenital neutropenia, bone marrow fibrosis, and extramedullary renal hematopoiesis. Detailed mechanisms of the VPS45 function are unknown. Here, we show an essential role of mammalian VPS45 in maintaining the intracellular organization of endolysosomal vesicles and promoting recycling of cell-surface receptors. Loss of VPS45 causes defective Rab5-to-Rab7 conversion resulting in trapping of cargos in early endosomes and impaired delivery to lysosomes. In this context, we demonstrate aberrant trafficking of the granulocyte colony-stimulating factor receptor in the absence of VPS45. Furthermore, we find that lack of VPS45 in mice is not compatible with embryonic development. Thus, we identify mammalian VPS45 as a critical regulator of trafficking through the endosomal system and early embryogenesis of mice.
- Published
- 2021
- Full Text
- View/download PDF
13. Rescue of STAT3 Function in Hyper-IgE Syndrome Using Adenine Base Editing
- Author
-
Michelle Plummer, Miriam T Kastlmeier, Renate Effner, Anna Pertek, Riccardo Berutti, Tobias Stoeger, Ellen D. Renner, Thomas Meitinger, Amina Sayed, Ejona Rusha, Florian Giesert, Thomas Volz, Beate Hagl, Carola Voss, Micha Drukker, Andreas C. Eberherr, Elisabeth Graf, Christine Wolf, Tim M. Strom, Andre Maaske, and Alena Buyx
- Subjects
STAT3 Transcription Factor ,Heterozygote ,Induced Pluripotent Stem Cells ,Immunoglobulin E ,Genetics ,Humans ,Medicine ,Clustered Regularly Interspaced Short Palindromic Repeats ,Base (exponentiation) ,STAT3 ,Gene Editing ,Whole Genome Sequencing ,biology ,business.industry ,Adenine ,High-Throughput Nucleotide Sequencing ,Cell Differentiation ,Fibroblasts ,medicine.disease ,Lung disease ,Mutation ,Immunology ,biology.protein ,Primary immunodeficiency ,CRISPR-Cas Systems ,business ,Job Syndrome ,Function (biology) ,Biotechnology - Abstract
STAT3-hyper IgE syndrome (STAT3-HIES) is a primary immunodeficiency presenting with destructive lung disease along with other symptoms. CRISPR-Cas9-mediated adenine base editors (ABEs) have the potential to correct one of the most common STAT3-HIES causing heterozygous
- Published
- 2021
- Full Text
- View/download PDF
14. c-Abl phosphorylation primes PARIS for neurodegeneration
- Author
-
Florian Giesert
- Subjects
ABL ,business.industry ,Neurodegeneration ,Cancer research ,medicine ,Phosphorylation ,Neurology (clinical) ,medicine.disease ,business - Abstract
This scientific commentary refers to ‘Parkin interacting substrate phosphorylation by c-Abl drives dopaminergic neurodegeneration’ by Kim et al. (doi:10.1093/brain/awab356).
- Published
- 2021
- Full Text
- View/download PDF
15. Identification of ocular regulatory functions of core histone variant H3.2
- Author
-
Sharmilee Vetrivel, Dong-Jiunn Jeffery Truong, Wolfgang Wurst, Jochen Graw, and Florian Giesert
- Subjects
Cellular and Molecular Neuroscience ,Ophthalmology ,Sensory Systems - Abstract
The posttranscriptional modifications (PTM) of the Histone H3 family play an important role in ocular system differentiation. However, there has been no study on the nature of specific Histone H3 subtype carrying these modifications. Fortuitously, we had previously identified a dominant small-eye mutant Aey69 mouse with a mutation in the H3.2 encoding Hist2h3c1 gene (Vetrivel et al., 2019). In continuation, in the present study, the role of Histone H3.2 with relation to the microphtalmic Aey69 has been elaborated. Foremost, a transgenic mouse line expressing the fusion protein H3.2-GFP was generated using Crispr/Cas9. The approach was intended to confer a unique tag to the Hist2h3c1 gene which is similar in sequence and encoded protein structure to other histones. The GFP tag was then used for ChIP Seq analysis of the genes regulated by H3.2. The approach revealed ocular specific H3.2 targets including Ephrin family genes. Altered enrichment of H3.2 was found in the mutant Aey69 mouse, specifically around the ligand Efna5 and the receptor Ephb2. The effect of this altered enrichment on Ephrin signaling was further analysed by QPCR and immunohistochemistry. This study identifies Hist2h3c1 encoded H3.2 as an important epigenetic player in ocular development. By binding to specific regions of ocular developmental factors Histone H3.2 facilitates the function of these genes for successful early ocular development.
- Published
- 2023
- Full Text
- View/download PDF
16. TRAF6 prevents fatal inflammation by homeostatic suppression of MALT1 protease
- Author
-
Oliver Plettenburg, Isabel Hamp, Thomas J. O’Neill, Carina Graß, Daniel Krappmann, Torben Gehring, Marie J. Tofaute, Andreas Gewies, Ronald Naumann, Katrin Demski, Henrik Schmidt, Martin Göttlicher, Florian Giesert, Marc Rosenbaum, Katharina Kriegsmann, Wolfgang Wurst, Thomas Seeholzer, Mark Kriegsmann, Theresa Schnalzger, Jürgen Ruland, Vigo Heissmeyer, and Tanja Poth
- Subjects
T cell ,Immunology ,Inflammation ,TRAF6 protein, mouse ,Biology ,medicine.disease_cause ,Malt1 protein, mouse ,Autoimmunity ,immunology [TNF Receptor-Associated Factor 6] ,Mice ,immunology [Inflammation] ,MALT1 protease ,immunology [Homeostasis] ,genetics [Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein] ,medicine ,Animals ,Homeostasis ,ddc:610 ,TNF Receptor-Associated Factor 6 ,General Medicine ,Acquired immune system ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein ,Genetically Engineered Mouse ,immunology [Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein] ,Female ,medicine.symptom ,genetics [TNF Receptor-Associated Factor 6] - Abstract
Balanced control of T cell signaling is critical for adaptive immunity and protection from autoimmunity. By combining genetically engineered mouse models, biochemical analyses and pharmacological interventions, we describe an unexpected dual role of the tumor necrosis factor receptor–associated factor 6 (TRAF6) E3 ligase as both a positive and negative regulator of mucosa-associated lymphoid tissue 1 (MALT1) paracaspase. Although MALT1-TRAF6 recruitment is indispensable for nuclear factor κB signaling in activated T cells, TRAF6 counteracts basal MALT1 protease activity in resting T cells. In mice, loss of TRAF6-mediated homeostatic suppression of MALT1 protease leads to severe autoimmune inflammation, which is completely reverted by genetic or therapeutic inactivation of MALT1 protease function. Thus, TRAF6 functions as a molecular brake for MALT1 protease in resting T cells and a signaling accelerator for MALT1 scaffolding in activated T cells, revealing that TRAF6 controls T cell activation in a switch-like manner. Our findings have important implications for development and treatment of autoimmune diseases.
- Published
- 2021
- Full Text
- View/download PDF
17. Disrupting Roquin-1 interaction with Regnase-1 induces autoimmunity and enhances antitumor responses
- Author
-
Stephanie L. Edelmann, Laura S. de Jonge, Lisa Kifinger, Wolfgang Wurst, Florian Giesert, Christine Hohn, Naoto Kawakami, Sebastian Theurich, Mingui Fu, Dierk Niessing, Nina Kronbeck, Martin E. Kirmaier, Vigo Heissmeyer, Timsse Raj, Thomas Monecke, Elena S. Davydova, Elaine H. Wong, Stefan Feske, Gesine Behrens, and Mariano Gonzalez Pisfil
- Subjects
Cytotoxicity, Immunologic ,Male ,Skin Neoplasms ,T-Lymphocytes ,Melanoma, Experimental ,Autoimmunity ,medicine.disease_cause ,Zc3h12a protein, mouse ,Immunotherapy, Adoptive ,immunology [T-Lymphocytes] ,Tumor Microenvironment ,Immunology and Allergy ,genetics [Ribonucleases] ,metabolism [Repressor Proteins] ,genetics [Ubiquitin-Protein Ligases] ,Mutation ,therapy [Skin Neoplasms] ,transplantation [T-Lymphocytes] ,metabolism [Skin Neoplasms] ,Cell biology ,medicine.anatomical_structure ,therapy [Melanoma, Experimental] ,Phenotype ,Female ,Protein Binding ,T cell ,Ubiquitin-Protein Ligases ,Immunology ,Mice, Transgenic ,Biology ,immunology [Melanoma, Experimental] ,Article ,Proinflammatory cytokine ,genetics [Skin Neoplasms] ,Immune system ,metabolism [Ubiquitin-Protein Ligases] ,Ribonucleases ,medicine ,Animals ,Humans ,ddc:610 ,metabolism [T-Lymphocytes] ,Autoantibody ,genetics [Melanoma, Experimental] ,Germinal center ,Immunity, Humoral ,Mice, Inbred C57BL ,Repressor Proteins ,genetics [Repressor Proteins] ,HEK293 Cells ,immunology [Skin Neoplasms] ,Rc3h1 protein, mouse ,metabolism [Melanoma, Experimental] ,metabolism [Ribonucleases] ,roquin-2 protein, mouse ,CD8 ,HeLa Cells - Abstract
Roquin and Regnase-1 proteins bind and post-transcriptionally regulate proinflammatory target messenger RNAs to maintain immune homeostasis. Either the sanroque mutation in Roquin-1 or loss of Regnase-1 cause systemic lupus erythematosus-like phenotypes. Analyzing mice with T cells that lack expression of Roquin-1, its paralog Roquin-2 and Regnase-1 proteins, we detect overlapping or unique phenotypes by comparing individual and combined inactivation. These comprised spontaneous activation, metabolic reprogramming and persistence of T cells leading to autoimmunity. Here, we define an interaction surface in Roquin-1 for binding to Regnase-1 that included the sanroque residue. Mutations in Roquin-1 impairing this interaction and cooperative regulation of targets induced T follicular helper cells, germinal center B cells and autoantibody formation. These mutations also improved the functionality of tumor-specific T cells by promoting their accumulation in the tumor and reducing expression of exhaustion markers. Our data reveal the physical interaction of Roquin-1 with Regnase-1 as a hub to control self-reactivity and effector functions in immune cell therapies. Mutations in the RNA-binding proteins Roquin-1 or Regnase-1 cause systemic autoimmunity. Heissmeyer and colleagues show that Roquin-1 and Regnase-1 physically interact and thereby regulate CD4+ and CD8+ T cell metabolism and functionality.
- Published
- 2021
- Full Text
- View/download PDF
18. Primary cilia and SHH signaling impairments in human and mouse models of Parkinson's disease
- Author
-
Sebastian Schmidt, Malte D. Luecken, Dietrich Trümbach, Sina Hembach, Kristina M. Niedermeier, Nicole Wenck, Klaus Pflügler, Constantin Stautner, Anika Böttcher, Heiko Lickert, Ciro Ramirez-Suastegui, Ruhel Ahmad, Michael J. Ziller, Julia C. Fitzgerald, Viktoria Ruf, Wilma D. J. van de Berg, Allert J. Jonker, Thomas Gasser, Beate Winner, Jürgen Winkler, Daniela M. Vogt Weisenhorn, Florian Giesert, Fabian J. Theis, Wolfgang Wurst, Anatomy and neurosciences, and Amsterdam Neuroscience - Neurodegeneration
- Subjects
Multidisciplinary ,metabolism [Parkinson Disease] ,General Physics and Astronomy ,Parkinson Disease ,metabolism [Neural Stem Cells] ,General Chemistry ,Shh protein, mouse ,genetics [Hedgehog Proteins] ,General Biochemistry, Genetics and Molecular Biology ,Disease Models, Animal ,Mice ,Neural Stem Cells ,genetics [Parkinson Disease] ,metabolism [Cilia] ,SHH protein, human ,Animals ,Humans ,Hedgehog Proteins ,ddc:500 ,metabolism [Hedgehog Proteins] ,Cilia ,Signal Transduction - Abstract
Parkinson’s disease (PD) as a progressive neurodegenerative disorder arises from multiple genetic and environmental factors. However, underlying pathological mechanisms remain poorly understood. Using multiplexed single-cell transcriptomics, we analyze human neural precursor cells (hNPCs) from sporadic PD (sPD) patients. Alterations in gene expression appear in pathways related to primary cilia (PC). Accordingly, in these hiPSC-derived hNPCs and neurons, we observe a shortening of PC. Additionally, we detect a shortening of PC in PINK1-deficient human cellular and mouse models of familial PD. Furthermore, in sPD models, the shortening of PC is accompanied by increased Sonic Hedgehog (SHH) signal transduction. Inhibition of this pathway rescues the alterations in PC morphology and mitochondrial dysfunction. Thus, increased SHH activity due to ciliary dysfunction may be required for the development of pathoetiological phenotypes observed in sPD like mitochondrial dysfunction. Inhibiting overactive SHH signaling may be a potential neuroprotective therapy for sPD.
- Published
- 2021
- Full Text
- View/download PDF
19. The Parkinson’s disease-linked Leucine-rich repeat kinase 2 (LRRK2) is required for insulin-stimulated translocation of GLUT4
- Author
-
Wolfgang Wurst, Thomas Ott, Kathrin Brockmann, Daniela Vogt-Weisenhorn, Andrea Wenninger-Weinzierl, Marita Munz, Ralf Kühn, Saskia Biskup, Florian Giesert, Natalja Funk, and Thomas Gasser
- Subjects
0301 basic medicine ,Cancer Research ,drug effects [Neuronal Outgrowth] ,metabolism [Leucine-Rich Repeat Serine-Threonine Protein Kinase-2] ,lcsh:Medicine ,pharmacology [Fibroblast Growth Factors] ,Mice ,0302 clinical medicine ,cytology [Fibroblasts] ,Insulin ,Phosphorylation ,lcsh:Science ,pharmacology [Insulin] ,Glucose Transporter Type 4 ,Multidisciplinary ,Kinase ,Parkinson Disease ,LRRK2 ,metabolism [rab GTP-Binding Proteins] ,Cell biology ,ddc ,Intracellular signal transduction ,metabolism [Proto-Oncogene Proteins c-akt] ,Signal transduction ,Technology Platforms ,metabolism [Fibroblasts] ,Signal Transduction ,drug effects [Signal Transduction] ,Cell Survival ,drug effects [Cell Survival] ,Neuronal Outgrowth ,metabolism [Parkinson Disease] ,Biology ,Leucine-rich repeat ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,Animals ,Humans ,Protein kinase B ,lcsh:R ,Fibroblasts ,metabolism [Glucose Transporter Type 4] ,pathology [Parkinson Disease] ,Rats ,nervous system diseases ,Fibroblast Growth Factors ,030104 developmental biology ,rab GTP-Binding Proteins ,biology.protein ,genetics [Leucine-Rich Repeat Serine-Threonine Protein Kinase-2] ,lcsh:Q ,Proto-Oncogene Proteins c-akt ,ddc:600 ,030217 neurology & neurosurgery ,GLUT4 - Abstract
Mutations within Leucine-rich repeat kinase 2 (LRRK2) are associated with late-onset Parkinson’s disease. The physiological function of LRRK2 and molecular mechanism underlying the pathogenic role of LRRK2 mutations remain uncertain. Here, we investigated the role of LRRK2 in intracellular signal transduction. We find that deficiency of Lrrk2 in rodents affects insulin-dependent translocation of glucose transporter type 4 (GLUT4). This deficit is restored during aging by prolonged insulin-dependent activation of protein kinase B (PKB, Akt) and Akt substrate of 160 kDa (AS160), and is compensated by elevated basal expression of GLUT4 on the cell surface. Furthermore, we find a crucial role of Rab10 phosphorylation by LRRK2 for efficient insulin signal transduction. Translating our findings into human cell lines, we find comparable molecular alterations in fibroblasts from Parkinson’s patients with the known pathogenic G2019S LRRK2 mutation. Our results highlight the role of LRRK2 in insulin-dependent signalling with potential therapeutic implications.
- Published
- 2019
- Full Text
- View/download PDF
20. Non-invasive and high-throughput interrogation of exon-specific isoform expression
- Author
-
Milica Živanić, Sigrid C. Schwarz, Tabea Strauß, Peter Heutink, Bianca Eßwein, Dominic Schwarz, Martin Zirngibl, Marcus Conrad, Christian Grätz, Francesco Leandro Vaccaro, Luisa Krumwiede, Julian Geilenkeuser, Wolfgang Wurst, Simone Göppert, Sebastian Doll, Florian Giesert, Christoph Gruber, Günter U. Höglinger, Tobias Santl, Gerald Raffl, Eva Magdalena Beck, Gil G. Westmeyer, Maren Beyer, Valentin Evsyukov, Dong-Jiunn Jeffery Truong, Enikő Baligács, Deniz Tümen, Johann Dietmar Körner, Niklas Armbrust, Teeradon Phlairaharn, and Eva-Maria Lederer
- Subjects
Gene isoform ,Proteomics ,CRISPR-Cas systems ,RNA splicing ,Proteome ,RNA Stability ,Induced Pluripotent Stem Cells ,MAPT protein, human ,tau Proteins ,Biology ,metabolism [RNA-Binding Proteins] ,metabolism [RNA, Messenger] ,Exon ,genetics [RNA, Messenger] ,Technical Report ,Protein splicing ,ddc:570 ,Humans ,Protein Isoforms ,FOXP1 protein, human ,genetics [RNA-Binding Proteins] ,RNA, Messenger ,Induced pluripotent stem cell ,Synthetic biology ,metabolism [Repressor Proteins] ,MBNL1 protein, human ,metabolism [Forkhead Transcription Factors] ,Alternative splicing ,Biological techniques ,High-throughput screening ,RNA-Binding Proteins ,Forkhead Transcription Factors ,Cell Biology ,FOXP1 ,Exons ,Embryonic stem cell ,metabolism [tau Proteins] ,Cell biology ,High-Throughput Screening Assays ,metabolism [Induced Pluripotent Stem Cells] ,Repressor Proteins ,Alternative Splicing ,genetics [Repressor Proteins] ,genetics [tau Proteins] ,HEK293 Cells ,genetics [Forkhead Transcription Factors] ,CRISPR-Cas Systems ,Single-Cell Analysis - Abstract
Expression of exon-specific isoforms from alternatively spliced mRNA is a fundamental mechanism that substantially expands the proteome of a cell. However, conventional methods to assess alternative splicing are either consumptive and work-intensive or do not quantify isoform expression longitudinally at the protein level. Here, we therefore developed an exon-specific isoform expression reporter system (EXSISERS), which non-invasively reports the translation of exon-containing isoforms of endogenous genes by scarlessly excising reporter proteins from the nascent polypeptide chain through highly efficient, intein-mediated protein splicing. We applied EXSISERS to quantify the inclusion of the disease-associated exon 10 in microtubule-associated protein tau (MAPT) in patient-derived induced pluripotent stem cells and screened Cas13-based RNA-targeting effectors for isoform specificity. We also coupled cell survival to the inclusion of exon 18b of FOXP1, which is involved in maintaining pluripotency of embryonic stem cells, and confirmed that MBNL1 is a dominant factor for exon 18b exclusion. EXSISERS enables non-disruptive and multimodal monitoring of exon-specific isoform expression with high sensitivity and cellular resolution, and empowers high-throughput screening of exon-specific therapeutic interventions., Truong et al. developed a cell-based reporter system, EXSISERS, that enables non-invasive quantification of the protein expression levels of exon-specific isoforms via intein-mediated protein splicing.
- Published
- 2021
- Full Text
- View/download PDF
21. Type 2 diabetes risk gene Dusp8 regulates hypothalamic Jnk signaling and insulin sensitivity
- Author
-
Florian Giesert, Dhiraj G. Kabra, Timo D. Müller, Kerstin Stemmer, Meri De Angelis, Martin Hrabé de Angelis, Fabian Seebacher, Martin Heni, Sonja C. Schriever, Ruchi Jain, Luke Harrison, Moya Wu, Martin Irmler, Rubén Nogueiras, Johannes Beckers, Hans-Ulrich Häring, Serge Luquet, Stephanie Kullmann, Chun-Xia Yi, Natalie Krahmer, Felipe Correa-da-Silva, Emily Violette Baumgart, Julien Castel, Paul T. Pfluger, Sarah Martinez, Peter Baumann, Mathias V. Schmidt, Jeffery D. Molkentin, Katrin Pfuhlmann, Hannah Schug, Jan Rozman, Wolfgang Wurst, Joachim Nagler, Matthias H. Tschöp, Helmholtz-Zentrum München (HZM), German Center for Diabetes Research - Deutsches Zentrum für Diabetesforschung [Neuherberg] (DZD), Technische Universität Munchen - Université Technique de Munich [Munich, Allemagne] (TUM), Institute for Diabetes Research and Metabolic Diseases [Tübingen, Germany], VU University Medical Center [Amsterdam], Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), TUM School of Life Sciences Weihenstephan, Skane University Hospital [Malmo], Lund University [Lund], Synlab [Lausanne], Unité de Biologie Fonctionnelle et Adaptative (BFA (UMR_8251 / U1133)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), School Life Science Weihenstephan (TUM), Eberhard Karls Universität Tübingen = Eberhard Karls University of Tuebingen, German Research Center for Neurodegenerative Diseases - Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich Cluster for systems neurology [Munich] (SyNergy), Technische Universität Munchen - Université Technique de Munich [Munich, Allemagne] (TUM)-Ludwig-Maximilians-Universität München (LMU), Institute of Molecular Genetics of the Czech Academy of Sciences (IMG / CAS), Czech Academy of Sciences [Prague] (CAS), Howard Hughes Medical Institute (HHMI), Cincinnati Children's Hospital Medical Center, Institute of the Royal Netherlands Academy of Arts and Sciences, Max Planck Institute of Psychiatry, Max-Planck-Gesellschaft, Endocrinology, Laboratory for Endocrinology, Amsterdam Neuroscience - Cellular & Molecular Mechanisms, and Amsterdam Gastroenterology Endocrinology Metabolism
- Subjects
0301 basic medicine ,MAP Kinase Kinase 4 ,[SDV]Life Sciences [q-bio] ,Type 2 diabetes ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Corticosterone ,enzymology [Hypothalamus] ,Glucose homeostasis ,genetics [Diabetes Mellitus, Experimental] ,Mice, Knockout ,Kinase ,Diabetes ,enzymology [Diabetes Mellitus, Experimental] ,General Medicine ,metabolism [Dual-Specificity Phosphatases] ,DUSP8 protein, mouse ,030220 oncology & carcinogenesis ,Dual-Specificity Phosphatases ,medicine.symptom ,Signal Transduction ,medicine.medical_specialty ,Hypothalamus ,enzymology [Diabetes Mellitus, Type 2] ,Inflammation ,genetics [Diabetes Mellitus, Type 2] ,Diabetes Mellitus, Experimental ,03 medical and health sciences ,Insulin resistance ,Commentaries ,Internal medicine ,Diabetes mellitus ,medicine ,Animals ,genetics [MAP Kinase Kinase 4] ,ddc:610 ,Obesity ,genetics [Dual-Specificity Phosphatases] ,business.industry ,metabolism [MAP Kinase Kinase 4] ,medicine.disease ,030104 developmental biology ,Endocrinology ,Metabolism ,Diabetes Mellitus, Type 2 ,chemistry ,Commentary ,Insulin Resistance ,business ,Hormone - Abstract
International audience; Recent genome-wide association studies (GWAS) identified DUSP8, encoding a dual-specificity phosphatase targeting mitogen-activated protein kinases, as a type 2 diabetes (T2D) risk gene. Here, we reveal that Dusp8 is a gatekeeper in the hypothalamic control of glucose homeostasis in mice and humans. Male, but not female, Dusp8 loss-of-function mice, either with global or corticotropin-releasing hormone neuron-specific deletion, had impaired systemic glucose tolerance and insulin sensitivity when exposed to high-fat diet (HFD). Mechanistically, we found impaired hypothalamic-pituitary-adrenal axis feedback, blunted sympathetic responsiveness, and chronically elevated corticosterone levels driven by hypothalamic hyperactivation of Jnk signaling. Accordingly, global Jnk1 ablation, AAV-mediated Dusp8 overexpression in the mediobasal hypothalamus, or metyrapone-induced chemical adrenalectomy rescued the impaired glucose homeostasis of obese male Dusp8-KO mice, respectively. The sex-specific role of murine Dusp8 in governing hypothalamic Jnk signaling, insulin sensitivity, and systemic glucose tolerance was consistent with functional MRI data in human volunteers that revealed an association of the DUSP8 rs2334499 risk variant with hypothalamic insulin resistance in men. Further, expression of DUSP8 was increased in the infundibular nucleus of T2D humans. In summary, our findings suggest the GWAS-identified gene Dusp8 as a novel hypothalamic factor that plays a functional role in the etiology of T2D.
- Published
- 2020
- Full Text
- View/download PDF
22. Determination of thyroid hormones in placenta using isotope-dilution liquid chromatography quadrupole time-of-flight mass spectrometry
- Author
-
Niels E. Skakkebæk, Meri De Angelis, Daniela Vogt-Weisenhorn, Ulla Feldt-Rasmussen, Karl-Werner Schramm, Hannu Kiviranta, Jorma Toppari, Katharina M. Main, Zhong-Min Li, Heqing Shen, and Florian Giesert
- Subjects
Thyroid Hormones ,Diiodothyronines ,Placenta ,Coefficient of variation ,Liquid-Liquid Extraction ,030209 endocrinology & metabolism ,Isotope dilution ,ta3111 ,Mass spectrometry ,01 natural sciences ,Biochemistry ,Mass Spectrometry ,Analytical Chemistry ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Isotopes ,Limit of Detection ,Pregnancy ,Animals ,Humans ,Solid phase extraction ,Detection limit ,Chromatography ,ta1184 ,Solid Phase Extraction ,010401 analytical chemistry ,Organic Chemistry ,Extraction (chemistry) ,Transplacental ,General Medicine ,Reference Standards ,0104 chemical sciences ,Thyroxine ,chemistry ,Isotope Labeling ,Thyronine ,Triiodothyronine ,Female ,Chromatography, Liquid - Abstract
The transplacental passage of thyroid hormones (THs) is of great significance since the maternal THs are vitally important in ensuring the normal fetal development. In this paper, we determined the concentrations of seven THs, viz. L-thyroxine (T4), 3,3′,5-triiodo- l -thyronine (T3), 3,3′,5′-triiodo- l -thyronine (rT3), 3,3′-diiodo- l -thyronine (T2), 3,5-diiodo- l -thyronine (rT2), 3-iodo- l -thyronine (T1) and 3-iodothyronamine (T1AM), in placenta using isotope dilution liquid chromatography quadrupole time-of-flight mass spectrometry. We optimized the method using isotopically labeled quantification standards (13C6-T4, 13C6-T3, 13C6-rT3 and 13C6-T2) and recovery standard (13C12-T4) in combination with solid-liquid extraction, liquid–liquid extraction and solid phase extraction. The linearity was obtained in the range of 0.5–150 pg uL−1 with R2 values >0.99. The method detection limits (MDLs) ranged from 0.01 ng g−1 to 0.2 ng g−1, while the method quantification limits (MQLs) were between 0.04 ng g−1 and 0.7 ng g−1. The spike-recoveries for THs (except for T1 and T1AM) were in the range of 81.0%–112%, with a coefficient of variation (CV) of 0.5–6.2%. The intra-day CVs and inter-day CVs were 0.5%–10.3% and 1.19%–8.88%, respectively. Concentrations of the THs were 22.9–35.0 ng g−1 T4, 0.32–0.46 ng g−1 T3, 2.86–3.69 ng g−1 rT3, 0.16–0.26 ng g−1 T2, and
- Published
- 2018
- Full Text
- View/download PDF
23. The pathogenic LRRK2 R1441C mutation induces specific deficits modeling the prodromal phase of Parkinson's disease in the mouse
- Author
-
Luise Ernst, D. M. Vogt Weisenhorn, Lisa Glasl, Annemarie Zimprich, Florian Giesert, G. Piccoli, Sabine M. Hölter, Julia Zerle, Wolfgang Wurst, and C. Stautner
- Subjects
0301 basic medicine ,Parkinson's disease ,Prodromal symptoms ,metabolism [Leucine-Rich Repeat Serine-Threonine Protein Kinase-2] ,Behavioral phenotyping ,medicine.disease_cause ,Mice ,0302 clinical medicine ,metabolism [Synaptotagmin I] ,genetics [Parkinson Disease] ,Neurodegenertion ,genetics [Point Mutation] ,psychology [Swimming] ,Gait ,genetics [Arginine] ,Genetics ,Mutation ,Gene knockdown ,Neurodegeneration ,Parkinson Disease ,LRRK2 ,Phenotype ,Smell ,Neurology ,Synaptotagmin I ,Genetic mouse model of Parkinson's disease ,physiopathology [Parkinson Disease] ,physiology [Recognition, Psychology] ,Genotype ,Tyrosine 3-Monooxygenase ,genetics [Smell] ,genetics [Cysteine] ,Synaptophysin ,Prodromal Symptoms ,Mice, Transgenic ,genetics [Motor Activity] ,Knock-out–knock-in models ,Motor Activity ,Biology ,Arginine ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,lcsh:RC321-571 ,03 medical and health sciences ,ddc:570 ,medicine ,Point Mutation ,Animals ,Cysteine ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Swimming ,Loss function ,Point mutation ,metabolism [Synaptophysin] ,Recognition, Psychology ,medicine.disease ,pathology [Parkinson Disease] ,metabolism [Tyrosine 3-Monooxygenase] ,nervous system diseases ,Disease Models, Animal ,030104 developmental biology ,Exploratory Behavior ,genetics [Leucine-Rich Repeat Serine-Threonine Protein Kinase-2] ,genetics [Gait] ,Neuroscience ,030217 neurology & neurosurgery ,physiology [Exploratory Behavior] - Abstract
The aim of the present study was to further explore the in vivo function of the Leucine-rich repeat kinase 2 (LRRK2)-gene, which is mutated in certain familial forms of Parkinson's disease (PD). We generated a mouse model harboring the disease-associated point mutation R1441C in the GTPase domain of the endogenous murine LRRK2 gene (LRRK2 R1441C line) and performed a comprehensive analysis of these animals throughout lifespan in comparison with an existing knockdown line of LRRK2 (LRRK2 knockdown line). Animals of both lines do not exhibit severe motor dysfunction or pathological signs of neurodegeneration neither at young nor old age. However, at old age the homozygous LRRK2 R1441C animals exhibit clear phenotypes related to the prodromal phase of PD such as impairments in fine motor tasks, gait, and olfaction. These phenotypes are only marginally observable in the LRRK2 knockdown animals, possibly due to activation of compensatory mechanisms as suggested by in vitro studies of synaptic transmission. Thus, at the organismal level the LRRK2 R1441C mutation does not emerge as a loss of function of the protein, but induces mutation specific deficits. Furthermore, judged by the phenotypes presented, the LRRK2-R1441C knock-in line is a valid preclinical model for the prodromal phase of PD.
- Published
- 2017
- Full Text
- View/download PDF
24. CRISPR-Mediated Induction of Neuron-Enriched Mitochondrial Proteins Boosts Direct Glia-to-Neuron Conversion
- Author
-
Giovanna Sonsalla, Christopher T. Breunig, Jessica Giehrl-Schwab, Magdalena Götz, Juliane Merl-Pham, Florian Giesert, Hans Zischka, Sabine Schmitt, Wolfgang Wurst, Gianluca Luigi Russo, Martin Jastroch, Stefan H. Stricker, Stefanie M. Hauck, Poornemaa Natarajan, Giacomo Masserdotti, and Giorgia Bulli
- Subjects
Cell type ,antioxidant ,proteome ,SOD1 ,Mitochondrion ,Biology ,Mitochondrial Proteins ,03 medical and health sciences ,Short Article ,0302 clinical medicine ,ddc:570 ,medicine ,Genetics ,CRISPR ,Clustered Regularly Interspaced Short Palindromic Repeats ,Cells, Cultured ,CRISPR-a ,030304 developmental biology ,Neurons ,0303 health sciences ,direct reprogramming ,metabolism ,mitochondria ,Correction ,Cell Biology ,Cell biology ,ddc ,medicine.anatomical_structure ,nervous system ,Astrocytes ,Proteome ,Molecular Medicine ,genetics [Mitochondrial Proteins] ,Neuron ,Reprogramming ,Neuroglia ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Summary Astrocyte-to-neuron conversion is a promising avenue for neuronal replacement therapy. Neurons are particularly dependent on mitochondrial function, but how well mitochondria adapt to the new fate is unknown. Here, we determined the comprehensive mitochondrial proteome of cortical astrocytes and neurons, identifying about 150 significantly enriched mitochondrial proteins for each cell type, including transporters, metabolic enzymes, and cell-type-specific antioxidants. Monitoring their transition during reprogramming revealed late and only partial adaptation to the neuronal identity. Early dCas9-mediated activation of genes encoding mitochondrial proteins significantly improved conversion efficiency, particularly for neuron-enriched but not astrocyte-enriched antioxidant proteins. For example, Sod1 not only improves the survival of the converted neurons but also elicits a faster conversion pace, indicating that mitochondrial proteins act as enablers and drivers in this process. Transcriptional engineering of mitochondrial proteins with other functions improved reprogramming as well, demonstrating a broader role of mitochondrial proteins during fate conversion., Graphical abstract, Highlights • Mitochondrial proteomes of cortical astrocytes and neurons are distinct • Astrocyte-enriched mitochondrial proteins are downregulated late in neuronal conversion • Neuron-enriched mitochondrial proteins are upregulated late in neuronal conversion • Early induction of neuronal mitochondrial proteins improves neuronal reprogramming, Russo et al. identify mitochondrial proteins enriched in neurons or astrocytes. Astrocyte-enriched mitochondrial proteins are often only partially downregulated during astrocyte-to-neuron direct reprogramming. Neuron-enriched ones are upregulated late and mainly in reprogrammed neurons. CRISPRa-mediated early induction of neuron-enriched mitochondrial proteins boosts direct neuronal reprogramming speed and efficiency.
- Published
- 2019
- Full Text
- View/download PDF
25. A validated LC-Q-TOF-MS method for quantitative analysis of thyroxine and metabolites in placenta
- Author
-
Heqing Shen, Niels E Skakkebaek, K M Main, Zhong-Min Li, Ulla Feldt-Rasmussen, Daniela Vogt-Weisenhorn, Karl-Werner Schramm, Meri De Angelis, Jorma Toppari, Florian Giesert, and Hannu Kiviranta
- Subjects
Lc q tof ms ,medicine.anatomical_structure ,Chromatography ,Chemistry ,Placenta ,medicine ,Quantitative analysis (chemistry) - Published
- 2018
- Full Text
- View/download PDF
26. Analysis of locomotor behavior in the German Mouse Clinic
- Author
-
Jan Rozman, Valerie Gailus-Durner, Angelika Hummel, Annemarie Zimprich, Petra Dirscherl, Manuela A. Östereicher, Lisa Glasl, Florian Giesert, Martin Hrabě de Angelis, Wolfgang Wurst, Lillian Garrett, Lore Becker, Sabine M. Hölter, Helmut Fuchs, Luise Ernst, and Daniela Vogt-Weisenhorn
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Genotype ,Mice, Transgenic ,Biology ,Body weight ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Physical medicine and rehabilitation ,Gait (human) ,physiology [Locomotion] ,medicine ,Animals ,ddc:610 ,Principal Component Analysis ,Behavior, Animal ,General Neuroscience ,Activity ,Catwalk ,Locomotion ,Mouse ,Phenotyping ,Data set ,physiology [Behavior, Animal] ,030104 developmental biology ,Phenotype ,methods [Behavioral Research] ,Gait analysis ,Principal component analysis ,Female ,Gait Analysis ,Neuroscience ,methods [Gait Analysis] ,030217 neurology & neurosurgery ,Behavioral Research - Abstract
Background Generation and phenotyping of mutant mouse models continues to increase along with the search for the most efficient phenotyping tests. Here we asked if a combination of different locomotor tests is necessary for comprehensive locomotor phenotyping, or if a large data set from an automated gait analysis with the CatWalk system would suffice. New method First we endeavored to meaningfully reduce the large CatWalk data set by Principal Component Analysis (PCA) to decide on the most relevant parameters. We analyzed the influence of sex, body weight, genetic background and age. Then a combination of different locomotor tests was analyzed to investigate the possibility of redundancy between tests. Result The extracted 10 components describe 80% of the total variance in the CatWalk, characterizing different aspects of gait. With these, effects of CatWalk version, sex, body weight, age and genetic background were detected. In addition, the PCA on a combination of locomotor tests suggests that these are independent without significant redundancy in their locomotor measures. Comparison with existing methods The PCA has permitted the refinement of the highly dimensional CatWalk (and other tests) data set for the extraction of individual component scores and subsequent analysis. Conclusion The outcome of the PCA suggests the possibility to focus on measures of the front and hind paws, and one measure of coordination in future experiments to detect phenotypic differences. Furthermore, although the CatWalk is sensitive for detecting locomotor phenotypes pertaining to gait, it is necessary to include other tests for comprehensive locomotor phenotyping.
- Published
- 2017
- Full Text
- View/download PDF
27. LRRK2 guides the actin cytoskeleton at growth cones together with ARHGEF7 and Tropomyosin 4
- Author
-
Karina Häbig, Thomas Hentrich, Birgit Heim, Florian Giesert, Carolin Walter, Olaf Riess, Michael Bonin, Wolfgang Wurst, Sandra Gellhaar, and Verena Djuric
- Subjects
genetics [Neuroblastoma] ,Parkinson's disease ,metabolism [Rho Guanine Nucleotide Exchange Factors] ,Fluorescent Antibody Technique ,Arp2/3 complex ,metabolism [Hippocampus] ,genetics [RNA, Small Interfering] ,Tropomyosin ,Hippocampus ,Immunoenzyme Techniques ,Mice ,Neuroblastoma ,antagonists & inhibitors [Rho Guanine Nucleotide Exchange Factors] ,pathology [Neuroblastoma] ,Guanine Nucleotide Exchange Factors ,metabolism [Actin Cytoskeleton] ,genetics [Tropomyosin] ,RNA, Small Interfering ,metabolism [Tropomyosin] ,TPM4 protein, human ,Cells, Cultured ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,Neurons ,Reverse Transcriptase Polymerase Chain Reaction ,Neurite outgrowth ,metabolism [Protein-Serine-Threonine Kinases] ,metabolism [Neurites] ,Protein-Serine-Threonine Kinases ,LRRK2 ,Cell biology ,Actin Cytoskeleton ,metabolism [Neurons] ,Molecular Medicine ,Lrrk2 protein, mouse ,antagonists & inhibitors [Protein-Serine-Threonine Kinases] ,metabolism [Biomarkers] ,Growth cone ,Neurite ,metabolism [Growth Cones] ,Blotting, Western ,Growth Cones ,metabolism [Neuroblastoma] ,macromolecular substances ,genetics [Rho Guanine Nucleotide Exchange Factors] ,Protein Serine-Threonine Kinases ,Biology ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Real-Time Polymerase Chain Reaction ,genetics [Protein-Serine-Threonine Kinases] ,Filamentous actin ,genetics [RNA, Messenger] ,Neurites ,Animals ,Humans ,ddc:610 ,LRRK2 protein, human ,RNA, Messenger ,Molecular Biology ,Actin ,Cell Proliferation ,physiology [Protein-Serine-Threonine Kinases] ,Gene Expression Profiling ,Actin cytoskeleton ,nervous system diseases ,ARHGEF7 protein, human ,cytology [Hippocampus] ,cytology [Neurons] ,NIH 3T3 Cells ,biology.protein ,Biomarkers ,Rho Guanine Nucleotide Exchange Factors - Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene represent the most common genetic cause of Parkinson's disease (PD). However, LRRK2 function and molecular mechanisms causing the parkinsonian phenotype remain widely unknown. Most of LRRK2 knockdown and overexpression models strengthen the relevance of LRRK2 in regulating neurite outgrowth. We have recently identified ARHGEF7 as the first guanine nucleotide exchange factor (GEF) of LRRK2. This GEF is influencing neurite outgrowth through regulation of actin polymerization. Here, we examined the expression profile of neuroblastoma cells with reduced LRRK2 and ARHGEF7 levels to identify additional partners of LRRK2 in this process. Tropomyosins (TPMs), and in particular TPM4, were the most interesting candidates next to other actin cytoskeleton regulating transcripts in this dataset. Subsequently, enhanced neurite branching was shown using primary hippocampal neurons of LRRK2 knockdown animals. Furthermore, we observed an enhanced number of growth cones per neuron and a mislocalization and dysregulation of ARHGEF7 and TPM4 in these neuronal compartments. Our results reveal a fascinating connection between the neurite outgrowth phenotype of LRRK2 models and the regulation of actin polymerization directing further investigations of LRRK2-related pathogenesis.
- Published
- 2013
- Full Text
- View/download PDF
28. Determination of thyroid hormones in small amount of mouse tissue sample using new isotope-dilution liquid chromatography-mass spectrometry method
- Author
-
Karl-Werner Schramm, Timo Dirk Mueller, Christoffer Clemmensen, Matthias Tschoep, Florian Giesert, Angelis Meri De, and Brian Finan
- Subjects
Chromatography ,Chemistry ,Liquid chromatography–mass spectrometry ,Sample (material) ,Thyroid hormones ,Analytical chemistry ,Mouse tissue ,Isotope dilution - Published
- 2016
- Full Text
- View/download PDF
29. Restless legs syndrome-associated intronic common variant in Meis1 alters enhancer function in the developing telencephalon
- Author
-
Helmut Fuchs, Miguel Torres, Lillian Garrett, Derek Spieler, Christine von Toerne, Darina Czamara, José Bessa, Melanie Waldenberger, Florian Giesert, Peter Lichtner, José Luis Gómez-Skarmeta, Melina Claussnitzer, Thomas Meitinger, Juliane Winkelmann, Helmut Laumen, Bertram Müller-Myhsok, Johannes Beckers, Sabine M. Hölter, Martin Klingenspor, Marion Horsch, Hyun-Ok Kate Lee, Martin Hrabé de Angelis, Wolfgang Wurst, Juan J. Tena, Barbara Schormair, Franziska Knauf, Jan Rozman, Valerie Gailus-Durner, Erik Tilch, Ronald Naumann, Fernando Casares, Christian Gieger, Nazanin Karbalai, Stefanie M. Hauck, Maria Kaffe, Fritz Thyssen Foundation, German Academic Exchange Service, Unión Europea. European Cooperation in Science and Technology (COST), Deutsche Forschungsgemeinschaft (Alemania), Helmholtz Zentrum München, Federal Ministry of Education & Research (Alemania), Ludwig-Maximilians-Universität München (Alemania), Ministerio de Ciencia e Innovación (España), Regional Government of Andalusia (España), Unión Europea. Fondo Europeo de Desarrollo Regional (FEDER/ERDF), Federal Ministry of Education and Research (Germany), German Research Center for Environmental Health, German Research Foundation, Israel Science Foundation, Technical University of Munich, Free State of Bavaria, Munich Center of Health Sciences, Ludwig Maximilians University Munich, Junta de Andalucía, German Center for Diabetes Research, and Helmholtz Association
- Subjects
Telencephalon ,genetics [Homeodomain Proteins] ,Single-nucleotide polymorphism ,Locus (genetics) ,Genome-wide association study ,Polymorphism, Single Nucleotide ,Basal Ganglia ,Mice ,pathology [Basal Ganglia] ,Restless Legs Syndrome ,mental disorders ,Genetics ,SNP ,Animals ,Allele ,Enhancer ,Myeloid Ecotropic Viral Integration Site 1 Protein ,Genetics (clinical) ,Alleles ,Homeodomain Proteins ,genetics [Neoplasm Proteins] ,biology ,genetics [Restless Legs Syndrome] ,Research ,metabolism [Basal Ganglia] ,growth & development [Telencephalon] ,Phenotype ,Introns ,Neoplasm Proteins ,Disease Models, Animal ,Enhancer Elements, Genetic ,pathology [Telencephalon] ,ddc:540 ,biology.protein ,CREB1 ,Meis1 protein, mouse ,Genome-Wide Association Study - Abstract
This article, published in Genome Research, is available under a Creative Commons License (Attribution-NonCommercial 3.0 Unported).-- et al., Genome-wide association studies (GWAS) identified the MEIS1 locus for Restless Legs Syndrome (RLS), but causal single nucleotide polymorphisms (SNPs) and their functional relevance remain unknown. This locus contains a large number of highly conserved noncoding regions (HCNRs) potentially functioning as cis-regulatory modules. We analyzed these HCNRs for allele-dependent enhancer activity in zebrafish and mice and found that the risk allele of the lead SNP rs12469063 reduces enhancer activity in the Meis1 expression domain of the murine embryonic ganglionic eminences (GE). CREB1 binds this enhancer and rs12469063 affects its binding in vitro. In addition, MEIS1 target genes suggest a role in the specification of neuronal progenitors in the GE, and heterozygous Meis1-deficient mice exhibit hyperactivity, resembling the RLS phenotype. Thus, in vivo and in vitro analysis of a common SNP with small effect size showed allele-dependent function in the prospective basal ganglia representing the first neurodevelopmental region implicated in RLS., The project was supported by Fritz-Thyssen-Stiftung, Cologne, Germany (10.09.2.146; 10.12.2.183), KKF-TUM (8766156), DAAD (0811963), and COST (“HOX and TALE homeoproteins in Development and Disease”). B.S. was partially supported by DFG grants (WI 1820/4-1; WI 1820/5-1) and a TUM-Excellence stipend. The KORA study was financed by the Helmholtz Zentrum München, which is funded by the German Federal Ministry of Education and Research (BMBF) and by the State of Bavaria. KORA research was supported within the Munich Center of Health Sciences (MC Health), Ludwig-Maximilians-Universität, as part of LMUinnovativ. J.L.G.-S. and F.C. acknowledge funding of the Spanish and the Andalusian Governments and the Feder program for grants (BFU2010-14839, BFU2009-07044, CSD2007-00008, and Proyectos de Excelencia CVI-3488 and CVI 2658). This work was funded in part by a grant from the German Federal Ministry of Education and Research (BMBF) to the German Center for Diabetes Research (DZD), to the German Mouse Clinic (Infrafrontier: 01KX1012), to the German Center for Neurodegenerative Diseases (DZNE), Germany; by the Initiative and Networking Fund of the Helmholtz Association in the framework of the Helmholtz Alliance for Mental Research in an Ageing Society (HA-215); and the Munich Cluster for Systems Neurology (EXC 1010 SyNergy) and its Collaborative Research Center (CRC) 870/2 “Assembly and Function of Neuronal Circuits.”
- Published
- 2014
- Full Text
- View/download PDF
30. Leucine-Rich Repeat Kinase 2 Binds to Neuronal Vesicles through Protein Interactions Mediated by Its C-Terminal WD40 Domain
- Author
-
Marius Ueffing, Franco Onofri, Michael Sattler, Christoph J. O. Kaiser, Andreas Kastenmüller, Michela Matteoli, Andreas Vogt, Christina Kiel, Maria Daniela Cirnaru, Francesca Pischedda, Mingjie Zhang, Pravin Kumar Ankush Jagtap, Carlo Sala, Christian Johannes Gloeckner, Lifeng Pan, Giovanni Piccoli, Flavia Antonucci, Sevil Weinkauf, Felix von Zweydorf, Florian Giesert, and Antonella Marte
- Subjects
Animals ,Cells, Cultured ,Humans ,Mice ,Mice, Inbred C57BL ,Mutant Proteins ,Mutation ,Neurons ,Neuropeptides ,Neurotoxins ,Parkinson Disease ,Protein Binding ,Protein Interaction Mapping ,Protein-Serine-Threonine Kinases ,Structure-Activity Relationship ,Synapses ,Synaptic Vesicles ,Protein Interaction Domains and Motifs ,Molecular Biology ,Cell Biology ,Cells ,Plasma protein binding ,Protein Serine-Threonine Kinases ,Biology ,Leucine-rich repeat ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Receptors for Activated C Kinase ,Inbred C57BL ,Synaptic vesicle ,Protein–protein interaction ,WD40 repeat ,Cultured ,Vesicle ,Articles ,LRRK2 ,nervous system diseases ,Cell biology ,Biochemistry ,Binding domain - Abstract
Mutations in the leucine-rich repeat kinase 2 gene (LRRK2) are associated with familial and sporadic Parkinson's disease (PD). LRRK2 is a complex protein that consists of multiple domains, including predicted C-terminal WD40 repeats. In this study, we analyzed functional and molecular features conferred by the WD40 domain. Electron microscopic analysis of the purified LRRK2 C-terminal domain revealed doughnut-shaped particles, providing experimental evidence for its WD40 fold. We demonstrate that LRRK2 WD40 binds and sequesters synaptic vesicles via interaction with vesicle-associated proteins. In fact, a domain-based pulldown approach combined with mass spectrometric analysis identified LRRK2 as being part of a highly specific protein network involved in synaptic vesicle trafficking. In addition, we found that a C-terminal sequence variant associated with an increased risk of developing PD, G2385R, correlates with a reduced binding affinity of LRRK2 WD40 to synaptic vesicles. Our data demonstrate a critical role of the WD40 domain within LRRK2 function.
- Published
- 2014
31. A cell surface biotinylation assay to reveal membrane-associated neuronal cues: Negr1 regulates dendritic arborization
- Author
-
Laura Cancedda, Marius Ueffing, Elena Vezzoli, Maria Daniela Cirnaru, Stefanie M. Hauck, Florian Giesert, Giovanni Piccoli, Carlo Sala, Joanna Szczurkowska, Francesca Pischedda, and Maura Francolini
- Subjects
Neurite ,Neuronal ,Cell Adhesion Molecules, Neuronal ,Cells ,Dendritic Spines ,Neurogenesis ,Biology ,Inbred C57BL ,Biochemistry ,Analytical Chemistry ,Cell membrane ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Humans ,Biotinylation ,Gene Silencing ,Cell Shape ,Molecular Biology ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,Cultured ,Neuronal growth regulator 1 ,Cell adhesion molecule ,Research ,Cell Membrane ,Membrane Proteins ,Cell Differentiation ,Dendrites ,Biological Assay ,HEK293 Cells ,Mice, Inbred C57BL ,Synapses ,Cell biology ,medicine.anatomical_structure ,Membrane protein ,Neuron maturation ,Immunoglobulin superfamily ,Cell Adhesion Molecules ,030217 neurology & neurosurgery - Abstract
A complex and still not comprehensively resolved panel of transmembrane proteins regulates the outgrowth and the subsequent morphological and functional development of neuronal processes. In order to gain a more detailed description of these events at the molecular level, we have developed a cell surface biotinylation assay to isolate, detect, and quantify neuronal membrane proteins. When we applied our assay to investigate neuron maturation in vitro, we identified 439 differentially expressed proteins, including 20 members of the immunoglobulin superfamily. Among these candidates, we focused on Negr1, a poorly described cell adhesion molecule. We demonstrated that Negr1 controls the development of neurite arborization in vitro and in vivo. Given the tight correlation existing among synaptic cell adhesion molecules, neuron maturation, and a number of neurological disorders, our assay results are a useful tool that can be used to support the understanding of the molecular bases of physiological and pathological brain function.
- Published
- 2014
32. FGF/FGFR2 signaling regulates the generation and correct positioning of Bergmann glia cells in the developing mouse cerebellum
- Author
-
Daniela M. Vogt Weisenhorn, Florian Meier, Sabit Delic, Theresa Faus-Kessler, Friederike Matheus, Ralf Kühn, Florian Giesert, Sabine M. Hölter, Nilima Prakash, Antonio Simeone, and Wolfgang Wurst
- Subjects
Cerebellum ,Cancer Research ,lcsh:Medicine ,Fibroblast growth factor ,Mice ,Cell Signaling ,Morphogenesis ,lcsh:Science ,Mice, Knockout ,Multidisciplinary ,cytology [Neuroglia] ,Cell migration ,Animal Models ,metabolism [Cerebellum] ,Cell biology ,Neuroepithelial cell ,metabolism [Receptor, Fibroblast Growth Factor, Type 2] ,Cell Motility ,cytology [Cerebellum] ,medicine.anatomical_structure ,Fibroblast growth factor receptor ,embryonic structures ,Neuroglia ,Technology Platforms ,Anatomy ,Signal Transduction ,Research Article ,Histology ,Cell Survival ,embryology [Cerebellum] ,Cell Migration ,Biology ,Research and Analysis Methods ,genetics [Receptor, Fibroblast Growth Factor, Type 2] ,Model Organisms ,FGF9 ,Developmental Neuroscience ,Neuroglial Development ,medicine ,Genetics ,Animals ,ddc:610 ,Receptor, Fibroblast Growth Factor, Type 2 ,metabolism [Neuroglia] ,Fgfr2 protein, mouse ,lcsh:R ,Biology and Life Sciences ,Cell Biology ,Molecular Development ,Embryonic stem cell ,metabolism [Fibroblast Growth Factors] ,Fibroblast Growth Factors ,Mice, Inbred C57BL ,stomatognathic diseases ,nervous system ,Mutagenesis ,Cellular Neuroscience ,Immunology ,Mutation ,lcsh:Q ,Gene Function ,Developmental Biology ,Neuroscience - Abstract
The normal cellular organization and layering of the vertebrate cerebellum is established during embryonic and early postnatal development by the interplay of a complex array of genetic and signaling pathways. Disruption of these processes and of the proper layering of the cerebellum usually leads to ataxic behaviors. Here, we analyzed the relative contribution of Fibroblast growth factor receptor 2 (FGFR2)-mediated signaling to cerebellar development in conditional Fgfr2 single mutant mice. We show that during embryonic mouse development, Fgfr2 expression is higher in the anterior cerebellar primordium and excluded from the proliferative ventricular neuroepithelium. Consistent with this finding, conditional Fgfr2 single mutant mice display the most prominent defects in the anterior lobules of the adult cerebellum. In this context, FGFR2-mediated signaling is required for the proper generation of Bergmann glia cells and the correct positioning of these cells within the Purkinje cell layer, and for cell survival in the developing cerebellar primordium. Using cerebellar microexplant cultures treated with an FGFR agonist (FGF9) or antagonist (SU5402), we also show that FGF9/FGFR-mediated signaling inhibits the outward migration of radial glia and Bergmann glia precursors and cells, and might thus act as a positioning cue for these cells. Altogether, our findings reveal the specific functions of the FGFR2-mediated signaling pathway in the generation and positioning of Bergmann glia cells during cerebellar development in the mouse.
- Published
- 2013
- Full Text
- View/download PDF
33. Pink1-deficiency in mice impairs gait, olfaction and serotonergic innervation of the olfactory bulb
- Author
-
Ralf Kühn, Ulrich Hafen, Martin Hrabé de Angelis, Andreas Hofmann, Karina Kloos, Daniela M. Vogt Weisenhorn, Julia Zerle, Lisa Glasl, Sabine M. Hölter, Konstanze F. Winklhofer, Florian Giesert, Anne Roethig, Jingzhong Zhang, Wolfgang Wurst, and Barbara Di Benedetto
- Subjects
Adrenergic Neurons ,physiology [Gait] ,PINK1 ,Cell Count ,Olfaction ,Mitochondrion ,Biology ,Motor Activity ,Serotonergic ,Mice ,Developmental Neuroscience ,metabolism [Serotonergic Neurons] ,medicine ,Animals ,ddc:610 ,metabolism [Protein Kinases] ,Gait ,metabolism [Olfactory Bulb] ,Kinase ,metabolism [Corpus Striatum] ,Dopaminergic Neurons ,metabolism [Dopaminergic Neurons] ,genetics [Protein Kinases] ,Neurodegeneration ,Dopaminergic ,physiology [Smell] ,metabolism [Mitochondria] ,medicine.disease ,Olfactory Bulb ,Corpus Striatum ,metabolism [Adrenergic Neurons] ,Olfactory bulb ,Mitochondria ,Smell ,Disease Models, Animal ,physiology [Motor Activity] ,Neurology ,PTEN-induced putative kinase ,Neuroscience ,Protein Kinases ,Serotonergic Neurons - Abstract
Parkinson's Disease (PD) is the most common neurodegenerative movement disorder. Autosomal-recessive mutations in the mitochondrial protein kinase PINK1 (PTEN-induced kinase 1) account for 1-2% of the hereditary early-onset cases. To study the mechanisms underlying disease development, we generated Pink1-deficient mice. In analogy to other genetic loss-of-function mouse models, Pink1(-/-) mice did not show morphological alterations in the dopaminergic system. As a consequence, no gross motor dysfunctions were observed indicating that these mice do not develop the cardinal symptoms of PD. Nonetheless, symptoms which develop mainly before bradykinesia, rigidity and resting tremor were clearly evident in Pink1-deficient mice. These symptoms were gait alterations and olfactory dysfunctions. Remarkably in the glomerular layer of the olfactory bulb the density of serotonergic fibers was significantly reduced. Concerning mitochondrial morphology, neurons in Pink1(-/-) mice had less fragmented mitochondria. In contrast, upon acute knock-down of Pink1 increased mitochondrial fragmentation was observed in neuronal cultures. This fragmentation was, however, evened out within days. Taken together, we demonstrate that Pink1-deficient mice exhibit behavioral symptoms of early phases of PD and present systematic experimental evidence for compensation of Pink1-deficiency at the cellular level. Thus, Pink1-deficient mice represent a model for the early phases of PD in which compensation may still impede the onset of neurodegeneration. Consequently, these mice are a valuable tool for studying Pink1-related PD development, as well as for searching for reliable PD biomarkers.
- Published
- 2011
- Full Text
- View/download PDF
34. LRRK2 controls synaptic vesicle storage and mobilization within the recycling pool
- Author
-
Giovanni Piccoli, Andrea Meixner, Matthias Bauer, Hakan Sarioglu, Karsten Boldt, Florian Giesert, Wolfgang Wurst, Carlo Sala, Christian Johannes Gloeckner, Silvia De Astis, Michela Matteoli, Daniela Vogt-Weisenhorn, Steven B. Condliffe, and Marius Ueffing
- Subjects
Patch-Clamp Techniques ,Postsynaptic Current ,Potassium Chloride ,Pathogenesis ,Mice ,0302 clinical medicine ,Tandem Mass Spectrometry ,RNA, Small Interfering ,N-Ethylmaleimide-Sensitive Proteins ,Cells, Cultured ,Cerebral Cortex ,Neurons ,0303 health sciences ,Kinase ,General Neuroscience ,Vesicle ,Gene Expression Regulation, Developmental ,LRRK2 ,Cell biology ,Protein Transport ,Synaptic Vesicles ,Sodium Channel Blockers ,Nerve Tissue Proteins ,Tetrodotoxin ,Biology ,Protein Serine-Threonine Kinases ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Transfection ,Synaptic vesicle ,Article ,03 medical and health sciences ,Microscopy, Electron, Transmission ,Gene silencing ,Animals ,Humans ,Immunoprecipitation ,030304 developmental biology ,Analysis of Variance ,READILY RELEASABLE POOL ,REPEAT KINASE 2 ,CLATHRIN-MEDIATED ENDOCYTOSIS ,CULTURED HIPPOCAMPAL-NEURONS ,SINGLE-PARTICLE TRACKING ,PAIRED-PULSE DEPRESSION ,MOTOR-NERVE TERMINALS ,PARKINSONS-DISEASE ,SHORT-TERM ,NEUROTRANSMITTER RELEASE ,Compartment (ship) ,Excitatory Postsynaptic Potentials ,Embryo, Mammalian ,Electric Stimulation ,nervous system diseases ,nervous system ,Mutation ,Synapses ,Calcium ,030217 neurology & neurosurgery ,Synaptosomes - Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the single most common cause of inherited Parkinson's disease. Little is known about its involvement in the pathogenesis of Parkinson's disease mainly because of the lack of knowledge about the physiological role of LRRK2. To determine the function of LRRK2, we studied the impact of short hairpin RNA-mediated silencing of LRRK2 expression in cortical neurons. Paired recording indicated that LRRK2 silencing affects evoked postsynaptic currents. Furthermore, LRRK2 silencing induces at the presynaptic site a redistribution of vesicles within the bouton, altered recycling dynamics, and increased vesicle kinetics. Accordingly, LRRK2 protein is present in the synaptosomal compartment of cortical neurons in which it interacts with several proteins involved in vesicular recycling. Our results suggest that LRRK2 modulates synaptic vesicle trafficking and distribution in neurons and in consequence participates in regulating the dynamics between vesicle pools inside the presynaptic bouton.
- Published
- 2011
35. DJ-1-deficient mice show less TH-positive neurons in the ventral tegmental area and exhibit non-motoric behavioural impairments
- Author
-
Holger Prokisch, Thomas Klopstock, Florian Giesert, Uwe Ahting, Thomas Floss, Sabine M. Hölter, Magdalena Kallnik, Philipp J. Kahle, Karin Görner, A. Röthig, M. Hrabé de Angelis, Klaus Beyer, Thu-Trang Pham, D. M. Vogt Weisenhorn, Karin Weindl, Wolfgang Wurst, and Lore Becker
- Subjects
Male ,Parkinson's disease ,Dopamine ,Protein Deglycase DJ-1 ,Striatum ,Behavioral Neuroscience ,Mice ,metabolism [Dopamine] ,Phosphorylation ,metabolism [Oncogene Proteins] ,Chromatography, High Pressure Liquid ,Neurons ,Mice, Knockout ,Oncogene Proteins ,Behavior, Animal ,Age Factors ,Immunohistochemistry ,Respiratory enzyme ,Mitochondria ,Up-Regulation ,Ventral tegmental area ,physiology [Behavior, Animal] ,medicine.anatomical_structure ,Neurology ,metabolism [Neurons] ,Knockout mouse ,genetics [Up-Regulation] ,Female ,metabolism [Mitogen-Activated Protein Kinase 8] ,genetics [Mitogen-Activated Protein Kinase 8] ,physiology [Recognition, Psychology] ,Genotype ,Tyrosine 3-Monooxygenase ,Blotting, Western ,genetics [Motor Activity] ,Motor Activity ,Biology ,Genetics ,medicine ,Animals ,Mitogen-Activated Protein Kinase 8 ,ddc:610 ,genetics [Phosphorylation] ,Loss function ,Analysis of Variance ,PARK7 protein, mouse ,Tyrosine hydroxylase ,Ventral Tegmental Area ,metabolism [Ventral Tegmental Area] ,PARK7 ,JNK Mitogen-Activated Protein Kinases ,Recognition, Psychology ,genetics [Oncogene Proteins] ,Peroxiredoxins ,medicine.disease ,metabolism [Mitochondria] ,metabolism [Tyrosine 3-Monooxygenase] ,genetics [JNK Mitogen-Activated Protein Kinases] ,metabolism [JNK Mitogen-Activated Protein Kinases] ,genetics [Mitochondria] ,Neuroscience - Abstract
Loss of function of DJ-1 (PARK7) is associated with autosomal recessive early-onset Parkinson's disease (PD), one of the major age-related neurological diseases. In this study, we extended former studies on DJ-1 knockout mice by identifying subtle morphological and behavioural phenotypes. The DJ-1 gene trap-induced null mutants exhibit less dopamine-producing neurons in the ventral tegmental area (VTA). They also exhibit slight changes in behaviour, i.e. diminished rearing behaviour and impairments in object recognition. Furthermore, we detected subtle phenotypes, which suggest that these animals compensate for the loss of DJ-1. First, we found a significant upregulation of mitochondrial respiratory enzyme activities, a mechanism known to protect against oxidative stress. Second, a close to significant increase in c-Jun N-terminal kinase 1 phosphorylation in old DJ-1-deficient mice hints at a differential activation of neuronal cell survival pathways. Third, as no change in the density of tyrosine hydroxylase (TH)-positive terminals in the striatum was observed, the remaining dopamine-producing neurons likely compensate by increasing axonal sprouting. In summary, the present data suggest that DJ-1 is implicated in major non-motor symptoms of PD appearing in the early phases of the disease-such as subtle impairments in motivated behaviour and cognition-and that under basal conditions the loss of DJ-1 is compensated.
- Published
- 2010
- Full Text
- View/download PDF
36. A powerful transgenic tool for fate mapping and functional analysis of newly generated neurons
- Author
-
Wolfgang Wurst, Jingzhong Zhang, Sebastien Couillard-Despres, Ludwig Aigner, Karina Kloos, Florian Giesert, and Daniela M. Vogt Weisenhorn
- Subjects
Doublecortin Domain Proteins ,metabolism [Stem Cells] ,Cre recombinase ,metabolism [Neural Stem Cells] ,pharmacology [Tamoxifen] ,genetics [Integrases] ,methods [Brain Mapping] ,Mice ,Neural Stem Cells ,genetics [Transgenes] ,genetics [Gene Expression Regulation, Developmental] ,Transgenes ,cytology [Neural Stem Cells] ,biosynthesis [Integrases] ,Brain Mapping ,General Neuroscience ,Stem Cells ,lcsh:QP351-495 ,Neurogenesis ,genetics [Cell Lineage] ,Gene Expression Regulation, Developmental ,Neural stem cell ,genetics [Neurogenesis] ,Microtubule-Associated Proteins ,Research Article ,genetics [Microtubule-Associated Proteins] ,genetics [Neuropeptides] ,Doublecortin Protein ,Transgene ,doublecortin protein ,Mice, Transgenic ,Biology ,drug effects [Gene Expression Regulation, Developmental] ,cytology [Stem Cells] ,lcsh:RC321-571 ,Cellular and Molecular Neuroscience ,Fate mapping ,Animals ,Cell Lineage ,ddc:610 ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Reporter gene ,Integrases ,Neuropeptides ,Doublecortin ,Mice, Inbred C57BL ,Tamoxifen ,lcsh:Neurophysiology and neuropsychology ,nervous system ,biology.protein ,NeuN ,Neuroscience - Abstract
Background Lack of appropriate tools and techniques to study fate and functional integration of newly generated neurons has so far hindered understanding of neurogenesis' relevance under physiological and pathological conditions. Current analyses are either dependent on mitotic labeling, for example BrdU-incorporation or retroviral infection, or on the detection of transient immature neuronal markers. Here, we report a transgenic mouse model (DCX-CreERT2) for time-resolved fate analysis of newly generated neurons. This model is based on the expression of a tamoxifen-inducible Cre recombinase under the control of a doublecortin (DCX) promoter, which is specific for immature neuronal cells in the CNS. Results In the DCX-CreERT2 transgenic mice, expression of CreERT2 was restricted to DCX+ cells. In the CNS of transgenic embryos and adult DCX-CreERT2 mice, tamoxifen administration caused the transient translocation of CreERT2 to the nucleus, allowing for the recombination of loxP-flanked sequences. In our system, tamoxifen administration at E14.5 resulted in reporter gene activation throughout the developing CNS of transgenic embryos. In the adult CNS, neurogenic regions were the primary sites of tamoxifen-induced reporter gene activation. In addition, reporter expression could also be detected outside of neurogenic regions in cells physiologically expressing DCX (e.g. piriform cortex, corpus callosum, hypothalamus). Four weeks after recombination, the vast majority of reporter-expressing cells were found to co-express NeuN, revealing the neuronal fate of DCX+ cells upon maturation. Conclusions This first validation demonstrates that our new DCX-CreERT2 transgenic mouse model constitutes a powerful tool to investigate neurogenesis, migration and their long-term fate of neuronal precursors. Moreover, it allows for a targeted activation or deletion of specific genes in neuronal precursors and will thereby contribute to unravel the molecular mechanisms controlling neurogenesis.
- Published
- 2010
- Full Text
- View/download PDF
37. Loss of DJ-1 impairs antioxidant response by altered glutamine and serine metabolism
- Author
-
Marcel Leist, Alessandro Michelucci, Florian Giesert, C. Stautner, D. M. Vogt Weisenhorn, Jenny Ghelfi, O. Popp, Christian Jäger, Lisa Krämer, Jochen G. Schneider, Stefan Schildknecht, Alexander Skupin, W. Wurst, Rejko Krüger, Johannes Meiser, Andre Wegner, A. Fouquier d'Herouel, Yannic Nonnenmacher, Daniel Weindl, Dirk Woitalla, Karsten Hiller, Xiangyi Dong, and Sylvie Delcambre
- Subjects
0301 basic medicine ,Parkinson's disease ,Glutamine ,Protein Deglycase DJ-1 ,genetics [Protein Deglycase DJ-1] ,Biochemistry, biophysics & molecular biology [F05] [Life sciences] ,Mitochondrion ,metabolism [Microglia] ,medicine.disease_cause ,Folate mediated one-carbon metabolism ,Antioxidants ,Serine ,chemistry.chemical_compound ,Mice ,Biochimie, biophysique & biologie moléculaire [F05] [Sciences du vivant] ,Cells, Cultured ,Neurons ,Microglia ,metabolism [Serine] ,GC/MS ,Dopaminergic ,ROS ,Glutathione ,Mitochondria ,3. Good health ,Cell biology ,metabolism [Protein Deglycase DJ-1] ,medicine.anatomical_structure ,Biochemistry ,Neurology ,metabolism [Neurons] ,Metabolome ,Stable isotope-assisted metabolomics ,Technology Platforms ,Biology ,lcsh:RC321-571 ,03 medical and health sciences ,Biosynthesis ,ddc:570 ,medicine ,Animals ,Humans ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,metabolism [Glutamine] ,PARK7 protein, mouse ,Oncogene ,metabolism [Mitochondria] ,Oxidative Stress ,030104 developmental biology ,chemistry ,Mitochondrial metabolism ,Oxidative stress ,metabolism [Antioxidants] - Abstract
The oncogene DJ-1 has been originally identified as a suppressor of PTEN. Further on, loss-of-function mutations have been described as a causative factor in Parkinson's disease (PD). DJ-1 has an important function in cellular antioxidant responses, but its role in central metabolism of neurons is still elusive. We applied stable isotope assisted metabolic profiling to investigate the effect of a functional loss of DJ-1 and show that DJ-1 deficient neuronal cells exhibit decreased glutamine influx and reduced serine biosynthesis. By providing precursors for GSH synthesis, these two metabolic pathways are important contributors to cellular antioxidant response. Down-regulation of these pathways, as a result of loss of DJ-1 leads to an impaired antioxidant response. Furthermore, DJ-1 deficient mouse microglia showed a weak but constitutive pro-inflammatory activation. The combined effects of altered central metabolism and constitutive activation of glia cells raise the susceptibility of dopaminergic neurons towards degeneration in patients harboring mutated DJ-1. Our work reveals metabolic alterations leading to increased cellular instability and identifies potential new intervention points that can further be studied in the light of novel translational medicine approaches. published
- Full Text
- View/download PDF
38. Human and mouse essentiality screens as a resource for disease gene discovery
- Author
-
Cacheiro, Pilar, Muñoz-Fuentes, Violeta, Westerberg, Henrik, Scott, R. H., Siddiq, A., Sieghart, A., Smith, K. R., Sosinsky, A., Spooner, W., Stevens, H. E., Stuckey, A., Sultana, R., Thomas, E. R. A., Konopka, Tomasz, Thompson, S. R., Tregidgo, C., Tucci, A., Walsh, E., Watters, S. A., Welland, M. J., Williams, E., Witkowska, K., Wood, S. M., Zarowiecki, M., Hsu, Chih-Wei, Marschall, Susan, Lengger, Christoph, Maier, Holger, Seisenberger, Claudia, Bürger, Antje, Kühn, Ralf, Schick, Joel, Hörlein, Andreas, Oritz, Oskar, Giesert, Florian, Christiansen, Audrey, Beig, Joachim, Kenyon, Janet, Codner, Gemma, Fray, Martin, Johnson, Sara J, Cleak, James, Szoke-Kovacs, Zsombor, Lafont, David, Vancollie, Valerie E, McLaren, Robbie S B, Lanza, Denise G, Hughes-Hallett, Lena, Rowley, Christine, Sanderson, Emma, Galli, Antonella, Tuck, Elizabeth, Green, Angela, Tudor, Catherine, Siragher, Emma, Dabrowska, Monika, Mazzeo, Cecilia Icoresi, Beaudet, Arthur L, Griffiths, Mark, Gannon, David, Doe, Brendan, Cockle, Nicola, Kirton, Andrea, Bottomley, Joanna, Ingle, Catherine, Ryder, Edward, Gleeson, Diane, Ramirez-Solis, Ramiro, Heaney, Jason D, Birling, Marie-Christine, Pavlovic, Guillaume, Ayadi, Abdel, Hamid, Meziane, About, Ghina Bou, Champy, Marie-France, Jacobs, Hugues, Wendling, Olivia, Leblanc, Sophie, Vasseur, Laurent, Fuchs, Helmut, Chesler, Elissa J, Kumar, Vivek, White, Jacqueline K, Svenson, Karen L, Wiegand, Jean-Paul, Anderson, Laura L, Wilcox, Troy, Clark, James, Ryan, Jennifer, Denegre, James, Gailus-Durner, Valerie, Stearns, Tim, Philip, Vivek, Witmeyer, Catherine, Bates, Lindsay, Seavey, Zachary, Stanley, Pamela, Willet, Amelia, Roper, Willson, Creed, Julie, Moore, Michayla, Sorg, Tania, Dorr, Alex, Fraungruber, Pamelia, Presby, Rose, Mckay, Matthew, Nguyen-Bresinsky, Dong, Goodwin, Leslie, Urban, Rachel, Kane, Coleen, Murray, Stephen A, Prochazka, Jan, Novosadova, Vendula, Lelliott, Christopher J, Wardle-Jones, Hannah, Wells, Sara, Teboul, Lydia, Cater, Heather, Stewart, Michelle, Hough, Tertius, Wurst, Wolfgang, Dickinson, Mary E, Sedlacek, Radislav, Adams, David J, Seavitt, John R, Tocchini-Valentini, Glauco, Mammano, Fabio, Braun, Robert E, McKerlie, Colin, Herault, Yann, de Angelis, Martin Hrabě, Mallon, Ann-Marie, Bucan, Maja, Lloyd, K C Kent, Brown, Steve D M, Parkinson, Helen, Meehan, Terrence F, Smedley, Damian, Consortium, Genomics England Research, Consortium, International Mouse Phenotyping, Ambrose, J. C., Arumugam, P., Baple, E. L., Nutter, Lauryl M J, Bleda, M., Boardman-Pretty, F., Boissiere, J. M., Boustred, C. R., Brittain, H., Caulfield, M. J., Chan, G. C., Craig, C. E. H., Daugherty, L. C., de Burca, A., Peterson, Kevin A, Devereau, A., Elgar, G., Foulger, R. E., Fowler, T., Furió-Tarí, P., Hackett, J. M., Halai, D., Hamblin, A., Henderson, S., Holman, J. E., Haselimashhadi, Hamed, Hubbard, T. J. P., Ibáñez, K., Jackson, R., Jones, L. J., Kasperaviciute, D., Kayikci, M., Lahnstein, L., Lawson, K., Leigh, S. E. A., Leong, I. U. S., Flenniken, Ann M, Lopez, F. J., Maleady-Crowe, F., Mason, J., McDonagh, E. M., Moutsianas, L., Mueller, M., Murugaesu, N., Need, A. C., Odhams, C. A., Patch, C., Morgan, Hugh, Perez-Gil, D., Polychronopoulos, D., Pullinger, J., Rahim, T., Rendon, A., Riesgo-Ferreiro, P., Rogers, T., Ryten, M., Savage, K., Sawant, K., Cacheiro, Pilar [0000-0002-6335-8208], Muñoz-Fuentes, Violeta [0000-0003-3574-546X], Nutter, Lauryl MJ [0000-0001-9619-146X], Peterson, Kevin A [0000-0001-8353-3694], Haselimashhadi, Hamed [0000-0001-7334-2421], Konopka, Tomasz [0000-0003-3042-4712], Hsu, Chih-Wei [0000-0002-9591-9567], Lanza, Denise G [0000-0001-8750-6933], Heaney, Jason D [0000-0001-8475-8828], Fuchs, Helmut [0000-0002-5143-2677], Gailus-Durner, Valerie [0000-0002-6076-0111], Lelliott, Christopher J [0000-0001-8087-4530], Adams, David J [0000-0001-9490-0306], Mammano, Fabio [0000-0003-3751-1691], McKerlie, Colin [0000-0002-2232-0967], Herault, Yann [0000-0001-7049-6900], de Angelis, Martin Hrabě [0000-0002-7898-2353], Lloyd, KC Kent [0000-0002-5318-4144], Smedley, Damian [0000-0002-5836-9850], Apollo - University of Cambridge Repository, Queen Mary University of London (QMUL), European Bioinformatics Institute [Hinxton] (EMBL-EBI), EMBL Heidelberg, The Jackson Laboratory [Bar Harbor] (JAX), Baylor College of Medicine (BCM), Baylor University, University of Pennsylvania, The Hospital for sick children [Toronto] (SickKids), Mount Sinai Hospital [Toronto, Canada] (MSH), MRC Harwell Institute [UK], Helmholtz Zentrum München = German Research Center for Environmental Health, Institut Clinique de la Souris (ICS), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), French National Infrastructure for Mouse Phenogenomics (PHENOMIN), Institute of Molecular Genetics of the Czech Academy of Sciences (IMG / CAS), Czech Academy of Sciences [Prague] (CAS), The Wellcome Trust Sanger Institute [Cambridge], Technische Universität München = Technical University of Munich (TUM), Ludwig-Maximilians-Universität München (LMU), CNR - Italian National Research Council (CNR), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), German Center for Diabetes Research - Deutsches Zentrum für Diabetesforschung [Neuherberg] (DZD), University of California [Davis] (UC Davis), University of California (UC), J C Ambrose, P Arumugam, E L Baple, M Bleda, F Boardman-Pretty, J M Boissiere, C R Boustred, H Brittain, M J Caulfield, G C Chan, C E H Craig, L C Daugherty, A de Burca, A Devereau, G Elgar, R E Foulger, T Fowler, P Furió-Tarí, J M Hackett, D Halai, A Hamblin, S Henderson, J E Holman, T J P Hubbard, K Ibáñez, R Jackson, L J Jones, D Kasperaviciute, M Kayikci, L Lahnstein, K Lawson, S E A Leigh, I U S Leong, F J Lopez, F Maleady-Crowe, J Mason, E M McDonagh, L Moutsianas, M Mueller, N Murugaesu, A C Need, C A Odhams, C Patch, D Perez-Gil, D Polychronopoulos, J Pullinger, T Rahim, A Rendon, P Riesgo-Ferreiro, T Rogers, M Ryten, K Savage, K Sawant, R H Scott, A Siddiq, A Sieghart, K R Smith, A Sosinsky, W Spooner, H E Stevens, A Stuckey, R Sultana, E R A Thomas, S R Thompson, C Tregidgo, A Tucci, E Walsh, S A Watters, M J Welland, E Williams, K Witkowska, S M Wood, M Zarowiecki, Susan Marschall, Christoph Lengger, Holger Maier, Claudia Seisenberger, Antje Bürger, Ralf Kühn, Joel Schick, Andreas Hörlein, Oskar Oritz, Florian Giesert, Joachim Beig, Janet Kenyon, Gemma Codner, Martin Fray, Sara J Johnson, James Cleak, Zsombor Szoke-Kovacs, David Lafont, Valerie E Vancollie, Robbie S B McLaren, Lena Hughes-Hallett, Christine Rowley, Emma Sanderson, Antonella Galli, Elizabeth Tuck, Angela Green, Catherine Tudor, Emma Siragher, Monika Dabrowska, Cecilia Icoresi Mazzeo, Mark Griffiths, David Gannon, Brendan Doe, Nicola Cockle, Andrea Kirton, Joanna Bottomley, Catherine Ingle, Edward Ryder, Diane Gleeson, Ramiro Ramirez-Solis, Marie-Christine Birling, Guillaume Pavlovic, Abdel Ayadi, Meziane Hamid, Ghina Bou About, Marie-France Champy, Hugues Jacobs, Olivia Wendling, Sophie Leblanc, Laurent Vasseur, Elissa J Chesler, Vivek Kumar, Jacqueline K White, Karen L Svenson, Jean-Paul Wiegand, Laura L Anderson, Troy Wilcox, James Clark, Jennifer Ryan, James Denegre, Tim Stearns, Vivek Philip, Catherine Witmeyer, Lindsay Bates, Zachary Seavey, Pamela Stanley, Amelia Willet, Willson Roper, Julie Creed, Michayla Moore, Alex Dorr, Pamelia Fraungruber, Rose Presby, Matthew Mckay, Dong Nguyen-Bresinsky, Leslie Goodwin, Rachel Urban, Coleen Kane, Herault, Yann, and Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)
- Subjects
0301 basic medicine ,Mutation rate ,Cancer Research ,[SDV]Life Sciences [q-bio] ,General Physics and Astronomy ,methods [Genetic Association Studies] ,Disease ,VARIANTS ,Mice ,Essential ,0302 clinical medicine ,IMPC ,Genetics research ,Lethal allele ,2.1 Biological and endogenous factors ,Aetiology ,lcsh:Science ,Organism ,ComputingMilieux_MISCELLANEOUS ,Disease gene ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,Genes, Essential ,genetics [Disease] ,Genomics ,R/BIOCONDUCTOR PACKAGE ,DATABASE ,UPDATE ,GENOME ,[SDV] Life Sciences [q-bio] ,Knockout mouse ,Identification (biology) ,ddc:500 ,International Mouse Phenotyping Consortium ,Technology Platforms ,Biotechnology ,Knockout ,Science ,Computational biology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Genetics ,medicine ,Animals ,Humans ,Genetic variation ,Clinical genetics ,Gene ,Genetic Association Studies ,030304 developmental biology ,Disease model ,Prevention ,Human Genome ,General Chemistry ,medicine.disease ,Developmental disorder ,Good Health and Well Being ,030104 developmental biology ,Genomics England Research Consortium ,Genes ,lcsh:Q ,Generic health relevance ,030217 neurology & neurosurgery ,Rare disease - Abstract
The identification of causal variants in sequencing studies remains a considerable challenge that can be partially addressed by new gene-specific knowledge. Here, we integrate measures of how essential a gene is to supporting life, as inferred from viability and phenotyping screens performed on knockout mice by the International Mouse Phenotyping Consortium and essentiality screens carried out on human cell lines. We propose a cross-species gene classification across the Full Spectrum of Intolerance to Loss-of-function (FUSIL) and demonstrate that genes in five mutually exclusive FUSIL categories have differing biological properties. Most notably, Mendelian disease genes, particularly those associated with developmental disorders, are highly overrepresented among genes non-essential for cell survival but required for organism development. After screening developmental disorder cases from three independent disease sequencing consortia, we identify potentially pathogenic variants in genes not previously associated with rare diseases. We therefore propose FUSIL as an efficient approach for disease gene discovery., Discovery of causal variants for monogenic disorders has been facilitated by whole exome and genome sequencing, but does not provide a diagnosis for all patients. Here, the authors propose a Full Spectrum of Intolerance to Loss-of-Function (FUSIL) categorization that integrates gene essentiality information to aid disease gene discovery.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.